Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Nature ; 545(7652): 108-111, 2017 05 04.
Article in English | MEDLINE | ID: mdl-28445460

ABSTRACT

Nine neurodegenerative diseases are caused by expanded polyglutamine (polyQ) tracts in different proteins, such as huntingtin in Huntington's disease and ataxin 3 in spinocerebellar ataxia type 3 (SCA3). Age at onset of disease decreases with increasing polyglutamine length in these proteins and the normal length also varies. PolyQ expansions drive pathogenesis in these diseases, as isolated polyQ tracts are toxic, and an N-terminal huntingtin fragment comprising exon 1, which occurs in vivo as a result of alternative splicing, causes toxicity. Although such mutant proteins are prone to aggregation, toxicity is also associated with soluble forms of the proteins. The function of the polyQ tracts in many normal cytoplasmic proteins is unclear. One such protein is the deubiquitinating enzyme ataxin 3 (refs 7, 8), which is widely expressed in the brain. Here we show that the polyQ domain enables wild-type ataxin 3 to interact with beclin 1, a key initiator of autophagy. This interaction allows the deubiquitinase activity of ataxin 3 to protect beclin 1 from proteasome-mediated degradation and thereby enables autophagy. Starvation-induced autophagy, which is regulated by beclin 1, was particularly inhibited in ataxin-3-depleted human cell lines and mouse primary neurons, and in vivo in mice. This activity of ataxin 3 and its polyQ-mediated interaction with beclin 1 was competed for by other soluble proteins with polyQ tracts in a length-dependent fashion. This competition resulted in impairment of starvation-induced autophagy in cells expressing mutant huntingtin exon 1, and this impairment was recapitulated in the brains of a mouse model of Huntington's disease and in cells from patients. A similar phenomenon was also seen with other polyQ disease proteins, including mutant ataxin 3 itself. Our data thus describe a specific function for a wild-type polyQ tract that is abrogated by a competing longer polyQ mutation in a disease protein, and identify a deleterious function of such mutations distinct from their propensity to aggregate.


Subject(s)
Ataxin-3/chemistry , Ataxin-3/metabolism , Autophagy , Beclin-1/metabolism , Peptides/metabolism , Animals , Ataxin-3/deficiency , Ataxin-3/genetics , Binding, Competitive , Brain/metabolism , Brain/pathology , Cell Line , Cells, Cultured , Disease Models, Animal , Exons/genetics , Female , Food Deprivation , Humans , Huntingtin Protein/chemistry , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Male , Mice , Mice, Inbred C57BL , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Neurons/cytology , Neurons/metabolism , Phagosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Domains , Protein Stability , Ubiquitin/metabolism
3.
Mol Cell ; 43(1): 19-32, 2011 Jul 08.
Article in English | MEDLINE | ID: mdl-21726807

ABSTRACT

Autophagy, a major degradation process for long-lived and aggregate-prone proteins, affects various human processes, such as development, immunity, cancer, and neurodegeneration. Several autophagy regulators have been identified in recent years. Here we show that nitric oxide (NO), a potent cellular messenger, inhibits autophagosome synthesis via a number of mechanisms. NO impairs autophagy by inhibiting the activity of S-nitrosylation substrates, JNK1 and IKKß. Inhibition of JNK1 by NO reduces Bcl-2 phosphorylation and increases the Bcl-2-Beclin 1 interaction, thereby disrupting hVps34/Beclin 1 complex formation. Additionally, NO inhibits IKKß and reduces AMPK phosphorylation, leading to mTORC1 activation via TSC2. Overexpression of nNOS, iNOS, or eNOS impairs autophagosome formation primarily via the JNK1-Bcl-2 pathway. Conversely, NOS inhibition enhances the clearance of autophagic substrates and reduces neurodegeneration in models of Huntington's disease. Our data suggest that nitrosative stress-mediated protein aggregation in neurodegenerative diseases may be, in part, due to autophagy inhibition.


Subject(s)
Autophagy , Nitric Oxide/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Beclin-1 , Cell Line , Class III Phosphatidylinositol 3-Kinases/metabolism , Enzyme Inhibitors/pharmacology , HEK293 Cells , HeLa Cells , Humans , Huntingtin Protein , Huntington Disease/metabolism , Huntington Disease/pathology , I-kappa B Kinase/metabolism , Mechanistic Target of Rapamycin Complex 1 , Membrane Proteins/metabolism , Mice , Mitogen-Activated Protein Kinase 8/metabolism , Multiprotein Complexes , NG-Nitroarginine Methyl Ester/pharmacology , Nerve Tissue Proteins/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Protein Isoforms/metabolism , Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , TOR Serine-Threonine Kinases , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
4.
Nat Chem Biol ; 11(5): 347-354, 2015 May.
Article in English | MEDLINE | ID: mdl-25848931

ABSTRACT

Huntington's disease (HD) is a currently incurable neurodegenerative condition caused by an abnormally expanded polyglutamine tract in huntingtin (HTT). We identified new modifiers of mutant HTT toxicity by performing a large-scale 'druggable genome' siRNA screen in human cultured cells, followed by hit validation in Drosophila. We focused on glutaminyl cyclase (QPCT), which had one of the strongest effects on mutant HTT-induced toxicity and aggregation in the cell-based siRNA screen and also rescued these phenotypes in Drosophila. We found that QPCT inhibition induced the levels of the molecular chaperone αB-crystallin and reduced the aggregation of diverse proteins. We generated new QPCT inhibitors using in silico methods followed by in vitro screening, which rescued the HD-related phenotypes in cell, Drosophila and zebrafish HD models. Our data reveal a new HD druggable target affecting mutant HTT aggregation and provide proof of principle for a discovery pipeline from druggable genome screen to drug development.


Subject(s)
Aminoacyltransferases/drug effects , Aminoacyltransferases/genetics , Huntington Disease/drug therapy , Huntington Disease/genetics , RNA, Small Interfering , Aminoacyltransferases/antagonists & inhibitors , Animals , Cells, Cultured , Computational Biology , Drosophila , Drug Evaluation, Preclinical , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Green Fluorescent Proteins/metabolism , Humans , Huntingtin Protein , Mice , Mice, Inbred C57BL , Mutation/genetics , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Zebrafish , alpha-Crystallin B Chain/metabolism
5.
Nat Genet ; 37(7): 771-6, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15980862

ABSTRACT

Mutations that affect the dynein motor machinery are sufficient to cause motor neuron disease. It is not known why there are aggregates or inclusions in affected tissues in mice with such mutations and in most forms of human motor neuron disease. Here we identify a new mechanism of inclusion formation by showing that decreased dynein function impairs autophagic clearance of aggregate-prone proteins. We show that mutations of the dynein machinery enhanced the toxicity of the mutation that causes Huntington disease in fly and mouse models. Furthermore, loss of dynein function resulted in premature aggregate formation by mutant huntingtin and increased levels of the autophagosome marker LC3-II in both cell culture and mouse models, compatible with impaired autophagosome-lysosome fusion.


Subject(s)
Adenine/analogs & derivatives , Autophagy , Dyneins/genetics , Huntington Disease/pathology , Mutation , Adenine/pharmacology , Adenylyl Imidodiphosphate/pharmacology , Animals , Behavior, Animal , Brain/pathology , COS Cells , Chlorocebus aethiops , Crosses, Genetic , Diptera , Dyneins/antagonists & inhibitors , Dyneins/metabolism , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Inclusion Bodies/metabolism , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , PC12 Cells , Proteasome Endopeptidase Complex/metabolism , Rats , Synucleins
6.
J Biol Chem ; 287(24): 20748-54, 2012 Jun 08.
Article in English | MEDLINE | ID: mdl-22461632

ABSTRACT

The generation and subsequent aggregation of amyloid ß (Aß) peptides play a crucial initiating role in the pathogenesis of Alzheimer disease (AD). The two main isoforms of these peptides have 40 (Aß(40)) or 42 residues (Aß(42)), the latter having a higher propensity to aggregate in vitro and being the main component of the plaques observed in vivo in AD patients. We have designed a series of tandem dimeric constructs of these Aß peptides to probe the manner in which changes in the aggregation kinetics of Aß affect its deposition and toxicity in a Drosophila melanogaster model system. The levels of insoluble aggregates were found to be substantially elevated in flies expressing the tandem constructs of both Aß(40) and Aß(42) compared with the equivalent monomeric peptides, consistent with the higher effective concentration, and hence increased aggregation rate, of the peptides in the tandem repeat. A unique feature of the Aß(42) constructs, however, is the appearance of high levels of soluble oligomeric aggregates and a corresponding dramatic increase in their in vivo toxicity. The toxic nature of the Aß(42) peptide in vivo can therefore be attributed to the higher kinetic stability of the oligomeric intermediate states that it populates relative to those of Aß(40) rather than simply to its higher rate of aggregation.


Subject(s)
Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Gene Expression , Peptide Fragments/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Disease Models, Animal , Drosophila melanogaster , Humans , Peptide Fragments/genetics , Protein Stability , Solubility
7.
Hum Mol Genet ; 19(23): 4573-86, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20829225

ABSTRACT

A major function of proteasomes and macroautophagy is to eliminate misfolded potentially toxic proteins. Mammalian proteasomes, however, cannot cleave polyglutamine (polyQ) sequences and seem to release polyQ-rich peptides. Puromycin-sensitive aminopeptidase (PSA) is the only cytosolic enzyme able to digest polyQ sequences. We tested whether PSA can protect against accumulation of polyQ fragments. In cultured cells, Drosophila and mouse muscles, PSA inhibition or knockdown increased aggregate content and toxicity of polyQ-expanded huntingtin exon 1. Conversely, PSA overexpression decreased aggregate content and toxicity. PSA inhibition also increased the levels of polyQ-expanded ataxin-3 as well as mutant α-synuclein and superoxide dismutase 1. These protective effects result from an unexpected ability of PSA to enhance macroautophagy. PSA overexpression increased, and PSA knockdown or inhibition reduced microtubule-associated protein 1 light chain 3-II (LC3-II) levels and the amount of protein degradation sensitive to inhibitors of lysosomal function and autophagy. Thus, by promoting autophagic protein clearance, PSA helps protect against accumulation of aggregation-prone proteins and proteotoxicity.


Subject(s)
Aminopeptidases/metabolism , Autophagy , Peptides/metabolism , Aminopeptidases/genetics , Animals , Ataxin-3 , Cell Line , Drosophila , Gene Knockdown Techniques , Humans , Huntingtin Protein , Male , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Peptide Fragments/metabolism , Proteasome Endopeptidase Complex/metabolism , RNA Interference , Repressor Proteins/genetics , Repressor Proteins/metabolism , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
8.
Hum Mol Genet ; 19(17): 3413-29, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20566712

ABSTRACT

Many neurodegenerative diseases exhibit protein accumulation and increased oxidative stress. Therapeutic strategies include clearing aggregate-prone proteins by enhancing autophagy or decreasing oxidative stress with antioxidants. Many autophagy-inducing stimuli increase reactive oxygen species (ROS), raising concerns that the benefits of autophagy up-regulation may be counterbalanced by ROS toxicity. Here we show that not all autophagy inducers significantly increase ROS. However, many antioxidants inhibit both basal and induced autophagy. By blocking autophagy, antioxidant drugs can increase the levels of aggregate-prone proteins associated with neurodegenerative disease. In fly and zebrafish models of Huntington's disease, antioxidants exacerbate the disease phenotype and abrogate the rescue seen with autophagy-inducing agents. Thus, the potential benefits in neurodegenerative diseases of some classes of antioxidants may be compromised by their autophagy-blocking properties.


Subject(s)
Antioxidants/administration & dosage , Autophagy/drug effects , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/physiopathology , Peptides/metabolism , Animals , COS Cells , Chlorocebus aethiops , Disease Models, Animal , Drosophila , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases/embryology , Neurodegenerative Diseases/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Zebrafish
9.
Nat Cell Biol ; 5(12): 1071-8, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14595335

ABSTRACT

The actin cytoskeleton is best known for its role during cellular morphogenesis. However, other evidence suggests that actin is also crucial for the organization and dynamics of membrane organelles such as endosomes and the Golgi complex. As in morphogenesis, the Rho family of small GTPases are key mediators of organelle actin-driven events, although it is unclear how these ubiquitously distributed proteins are activated to regulate actin dynamics in an organelle-specific manner. Here we show that the brain-specific Rho-binding protein Citron-N is enriched at, and associates with, the Golgi apparatus of hippocampal neurons in culture. Suppression of the whole protein or expression of a mutant form lacking the Rho-binding activity results in dispersion of the Golgi apparatus. In contrast, high intracellular levels induce localized accumulation of RhoA and filamentous actin, protecting the Golgi from the rupture normally produced by actin depolymerization. Biochemical and functional analyses indicate that Citron-N controls actin locally by assembling together the Rho effector ROCK-II and the actin-binding, neuron-specific, protein Profilin-IIa (PIIa). Together with recent data on endosomal dynamics, our results highlight the importance of organelle-specific Rho modulators for actin-dependent organelle organization and dynamics.


Subject(s)
Actin Cytoskeleton/metabolism , Cell Cycle Proteins , Cell Differentiation/physiology , Contractile Proteins , Golgi Apparatus/metabolism , Neurons/metabolism , Proteins/metabolism , rho GTP-Binding Proteins/metabolism , Actins/metabolism , Animals , Binding Sites/genetics , Cells, Cultured , Fetus , Hippocampus/metabolism , Intracellular Signaling Peptides and Proteins , Microfilament Proteins/metabolism , Neurons/ultrastructure , Profilins , Protein Binding/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/metabolism , Proteins/genetics , Rats , rho-Associated Kinases , rhoA GTP-Binding Protein/metabolism
10.
Hum Mol Genet ; 17(2): 170-8, 2008 Jan 15.
Article in English | MEDLINE | ID: mdl-17921520

ABSTRACT

Huntington's disease (HD) is caused by a polyglutamine expansion mutation in the huntingtin protein that confers a toxic gain-of-function and causes the protein to become aggregate-prone. Aggregate-prone proteins are cleared by macroautophagy, and upregulating this process by rapamycin, which inhibits the mammalian target of rapamycin (mTOR), attenuates their toxicity in various HD models. Recently, we demonstrated that lithium induces mTOR-independent autophagy by inhibiting inositol monophosphatase (IMPase) and reducing inositol and IP3 levels. Here we show that glycogen synthase kinase-3beta (GSK-3beta), another enzyme inhibited by lithium, has opposite effects. In contrast to IMPase inhibition that enhances autophagy, GSK3beta inhibition attenuates autophagy and mutant huntingtin clearance by activating mTOR. In order to counteract the autophagy inhibitory effects of mTOR activation resulting from lithium treatment, we have used the mTOR inhibitor rapamycin in combination with lithium. This combination enhances macroautophagy by mTOR-independent (IMPase inhibition by lithium) and mTOR-dependent (mTOR inhibition by rapamycin) pathways. We provide proof-of-principle for this rational combination treatment approach in vivo by showing greater protection against neurodegeneration in an HD fly model with TOR inhibition and lithium, or in HD flies treated with rapamycin and lithium, compared with either pathway alone.


Subject(s)
Autophagy/drug effects , Drosophila , Huntington Disease/drug therapy , Lithium Compounds/pharmacology , Sirolimus/pharmacology , Animals , COS Cells , Chlorocebus aethiops , Disease Models, Animal , Drosophila Proteins/antagonists & inhibitors , Female , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3 beta , Humans , Inositol/biosynthesis , Lithium Compounds/therapeutic use , Male , Mice , Phosphoinositide-3 Kinase Inhibitors , Protein Kinases , Sirolimus/therapeutic use , TOR Serine-Threonine Kinases
11.
J Cell Biol ; 170(7): 1101-11, 2005 Sep 26.
Article in English | MEDLINE | ID: mdl-16186256

ABSTRACT

Macroautophagy is a key pathway for the clearance of aggregate-prone cytosolic proteins. Currently, the only suitable pharmacologic strategy for up-regulating autophagy in mammalian cells is to use rapamycin, which inhibits the mammalian target of rapamycin (mTOR), a negative regulator of autophagy. Here we describe a novel mTOR-independent pathway that regulates autophagy. We show that lithium induces autophagy, and thereby, enhances the clearance of autophagy substrates, like mutant huntingtin and alpha-synucleins. This effect is not mediated by glycogen synthase kinase 3beta inhibition. The autophagy-enhancing properties of lithium were mediated by inhibition of inositol monophosphatase and led to free inositol depletion. This, in turn, decreased myo-inositol-1,4,5-triphosphate (IP3) levels. Our data suggest that the autophagy effect is mediated at the level of (or downstream of) lowered IP3, because it was abrogated by pharmacologic treatments that increased IP3. This novel pharmacologic strategy for autophagy induction is independent of mTOR, and may help treatment of neurodegenerative diseases, like Huntington's disease, where the toxic protein is an autophagy substrate.


Subject(s)
Autophagy/drug effects , Enzyme Inhibitors/pharmacology , Lithium/pharmacology , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/metabolism , Animals , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Humans , Huntingtin Protein , Inositol/metabolism , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Kinases/metabolism , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , alpha-Synuclein/genetics , alpha-Synuclein/metabolism
12.
Biochem J ; 412(2): 191-209, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18466116

ABSTRACT

Huntington's disease (HD) is a devastating autosomal dominant neurodegenerative disease caused by a CAG trinucleotide repeat expansion encoding an abnormally long polyglutamine tract in the huntingtin protein. Much has been learnt since the mutation was identified in 1993. We review the functions of wild-type huntingtin. Mutant huntingtin may cause toxicity via a range of different mechanisms. The primary consequence of the mutation is to confer a toxic gain of function on the mutant protein and this may be modified by certain normal activities that are impaired by the mutation. It is likely that the toxicity of mutant huntingtin is revealed after a series of cleavage events leading to the production of N-terminal huntingtin fragment(s) containing the expanded polyglutamine tract. Although aggregation of the mutant protein is a hallmark of the disease, the role of aggregation is complex and the arguments for protective roles of inclusions are discussed. Mutant huntingtin may mediate some of its toxicity in the nucleus by perturbing specific transcriptional pathways. HD may also inhibit mitochondrial function and proteasome activity. Importantly, not all of the effects of mutant huntingtin may be cell-autonomous, and it is possible that abnormalities in neighbouring neurons and glia may also have an impact on connected cells. It is likely that there is still much to learn about mutant huntingtin toxicity, and important insights have already come and may still come from chemical and genetic screens. Importantly, basic biological studies in HD have led to numerous potential therapeutic strategies.


Subject(s)
Huntington Disease , Mutation , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Trinucleotide Repeat Expansion , Animals , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/pathology , Huntington Disease/therapy , Mitochondria/metabolism , Models, Molecular , Nerve Tissue Proteins/chemistry , Nuclear Proteins/chemistry , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary , Reactive Oxygen Species/metabolism , Transcription, Genetic , Ubiquitin/metabolism
13.
Mol Biol Cell ; 15(11): 5053-63, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15371536

ABSTRACT

The mechanisms underlying completion of cytokinesis are still poorly understood. Here, we show that the Drosophila orthologue of mammalian Citron kinases is essential for the final events of the cytokinetic process. Flies bearing mutations in the Drosophila citron kinase (dck) gene were defective in both neuroblast and spermatocyte cytokinesis. In both cell types, early cytokinetic events such as central spindle assembly and contractile ring formation were completely normal. Moreover, cytokinetic rings constricted normally, leading to complete furrow ingression. However late telophases of both cell types displayed persistent midbodies associated with disorganized F actin and anillin structures. Similar defects were observed in dck RNA interference (RNAi) telophases, which, in addition to abnormal F actin and anillin rings, also displayed aberrant membrane protrusions at the cleavage site. Together, these results indicate that mutations in the dck gene result in morphologically abnormal intercellular bridges and in delayed resolution of these structures, suggesting that the wild-type function of dck is required for abscission at the end of cytokinesis. The phenotype of Dck-depleted cells is different from those observed in most Drosophila cytokinesis mutants but extraordinarily similar to that caused by anillin RNAi, suggesting that Dck and anillin are in the same pathway for completion of cytokinesis.


Subject(s)
Cytokinesis , Protein Serine-Threonine Kinases/physiology , Spermatocytes/cytology , Amino Acid Sequence , Animals , Blotting, Western , Cell Line , Cell Membrane/metabolism , DNA/metabolism , Drosophila melanogaster , Intracellular Signaling Peptides and Proteins , Male , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Neurons/metabolism , Phenotype , Phylogeny , Protein Serine-Threonine Kinases/genetics , RNA Interference , Sequence Homology, Amino Acid , Telophase
14.
Autophagy ; 13(9): 1613-1614, 2017 Sep 02.
Article in English | MEDLINE | ID: mdl-28722507

ABSTRACT

Expansions of polyglutamine (polyQ) tracts in different proteins cause 9 neurodegenerative conditions, such as Huntington disease and various ataxias. However, many normal mammalian proteins contain shorter polyQ tracts. As these are frequently conserved in multiple species, it is likely that some of these polyQ tracts have important but unknown biological functions. Here we review our recent study showing that the polyQ domain of the deubiquitinase ATXN3/ataxin-3 enables its interaction with BECN1/beclin 1, a key macroautophagy/autophagy initiator. ATXN3 regulates autophagy by deubiquitinating BECN1 and protecting it from proteasomal degradation. Interestingly, expanded polyQ tracts in other polyglutamine disease proteins compete with the shorter ATXN3 polyQ stretch and interfere with the ATXN3-BECN1 interaction. This competition results in decreased BECN1 levels and impaired starvation-induced autophagy, which phenocopies the loss of autophagic function mediated by ATXN3. Our findings describe a new autophagy-protective mechanism that may be altered in multiple neurodegenerative diseases.


Subject(s)
Autophagy/drug effects , Peptides/pharmacology , Animals , Ataxin-3/chemistry , Ataxin-3/metabolism , Humans , Models, Biological , Mutant Proteins/metabolism , Polymorphism, Genetic , Trinucleotide Repeat Expansion/genetics
15.
Nat Commun ; 7: 13821, 2016 12 08.
Article in English | MEDLINE | ID: mdl-27929117

ABSTRACT

Aberrant protein aggregation is controlled by various chaperones, including CCT (chaperonin containing TCP-1)/TCP-1/TRiC. Mutated CCT4/5 subunits cause sensory neuropathy and CCT5 expression is decreased in Alzheimer's disease. Here, we show that CCT integrity is essential for autophagosome degradation in cells or Drosophila and this phenomenon is orchestrated by the actin cytoskeleton. When autophagic flux is reduced by compromise of individual CCT subunits, various disease-relevant autophagy substrates accumulate and aggregate. The aggregation of proteins like mutant huntingtin, ATXN3 or p62 after CCT2/5/7 depletion is predominantly autophagy dependent, and does not further increase with CCT knockdown in autophagy-defective cells/organisms, implying surprisingly that the effect of loss-of-CCT activity on mutant ATXN3 or huntingtin oligomerization/aggregation is primarily a consequence of autophagy inhibition rather than loss of physiological anti-aggregation activity for these proteins. Thus, our findings reveal an essential partnership between two key components of the proteostasis network and implicate autophagy defects in diseases with compromised CCT complex activity.


Subject(s)
Autophagosomes/metabolism , Autophagy , Chaperonin Containing TCP-1/metabolism , Huntingtin Protein/metabolism , Protein Aggregation, Pathological/metabolism , Animals , Ataxin-3/metabolism , Drosophila , Female , HeLa Cells , Humans , Lysosomes/metabolism , Male , Mice, Transgenic , RNA-Binding Proteins/metabolism
16.
Brain Res Bull ; 60(4): 319-27, 2003 May 30.
Article in English | MEDLINE | ID: mdl-12781320

ABSTRACT

Small GTPases of the rho family regulate the extensive rearrangements of the cytoskeleton that characterize neuronal differentiation. Citron kinase is a target molecule for activated rhoA, previously implicated in control of cytokinesis. We have found that, in addition, it could play an important role in modulating the extension of neuronal processes. Using constitutively active and dominant negative mutants, we showed that citron kinase is involved in the morphologic differentiation of N1E-115 neuroblastoma cells induced by serum starvation. More importantly, quantitative analysis of citron kinase knockout cerebral cortex displayed that this molecule may differentially regulate the morphology of the dendritic compartment in corticocollicular versus callosally-projecting pyramidal neurons.


Subject(s)
Cerebral Cortex/enzymology , Dendrites/enzymology , Dendrites/physiology , Neurons/enzymology , Neurons/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Cell Differentiation/physiology , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Dendrites/ultrastructure , Gene Expression Regulation, Developmental/physiology , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Neurons/cytology , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Tumor Cells, Cultured
17.
Anticancer Res ; 24(5A): 2643-7, 2004.
Article in English | MEDLINE | ID: mdl-15517868

ABSTRACT

BACKGROUND: PIP3, generated by P13-K activates Akt which inactivates AFX/FKHR; with the consequent decrease in p27/Kip.1 expression and enhancement of cyclin D1 expression through FRAP/mTOR. PTEN lipid phosphatase degrades PIP3 and negatively regulates Akt, whereas this is activated by EGFR through PI3. In glioblastomas, PTEN is mutated in 27%-40% and EGFR amplified in 60%-65% of cases. MATERIALS AND METHODS: PTEN mutation and EGFR amplification by PCRP Akt, p27/Kip.1 and cyclin D1 by immunohistochemistry, apoptosis by TUNEL and MIB.1 LI were studied in a series of 65 operated glioblastomas. RESULTS: EGFR amplification and PTEN mutation were present in 50% and 30% of glioblastomas, respectively. No relationship between EGFR amplification and PTEN mutation, and p27/Kip. 1 and cyclin D1 was found. However, cyclin D1 was positive in 69% of Akt-expressing areas, whereas p27 was positive in 30% only. CONCLUSION: A direct relationship is more evident between cyclin D1 and p27/Kip.1 and Akt than with PTEN and EGFR.


Subject(s)
Cell Cycle Proteins/biosynthesis , Central Nervous System Neoplasms/metabolism , Cyclin D1/biosynthesis , Genes, erbB-1/genetics , Glioblastoma/metabolism , Phosphoric Monoester Hydrolases/genetics , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics , Central Nervous System Neoplasms/enzymology , Central Nervous System Neoplasms/genetics , Cyclin-Dependent Kinase Inhibitor p27 , ErbB Receptors/biosynthesis , ErbB Receptors/genetics , Gene Amplification , Glioblastoma/enzymology , Glioblastoma/genetics , Humans , Immunohistochemistry , Mutation , PTEN Phosphohydrolase , Proto-Oncogene Proteins c-akt
18.
Nat Commun ; 5: 4998, 2014 Sep 22.
Article in English | MEDLINE | ID: mdl-25241929

ABSTRACT

Genome-wide association studies have identified several loci associated with Alzheimer's disease (AD), including proteins involved in endocytic trafficking such as PICALM/CALM (phosphatidylinositol binding clathrin assembly protein). It is unclear how these loci may contribute to AD pathology. Here we show that CALM modulates autophagy and alters clearance of tau, a protein which is a known autophagy substrate and which is causatively linked to AD, both in vitro and in vivo. Furthermore, altered CALM expression exacerbates tau-mediated toxicity in zebrafish transgenic models. CALM influences autophagy by regulating the endocytosis of SNAREs, such as VAMP2, VAMP3 and VAMP8, which have diverse effects on different stages of the autophagy pathway, from autophagosome formation to autophagosome degradation. This study suggests that the AD genetic risk factor CALM modulates autophagy, and this may affect disease in a number of ways including modulation of tau turnover.


Subject(s)
Autophagy , Monomeric Clathrin Assembly Proteins/metabolism , tau Proteins/metabolism , Animals , Autophagy-Related Protein 12 , Cell Line , Drosophila , Endocytosis , Female , Fibroblasts/metabolism , Genome-Wide Association Study , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Phagosomes , Protein Binding , RNA, Small Interfering/metabolism , Risk Factors , Small Ubiquitin-Related Modifier Proteins/metabolism , Transfection , Vesicle-Associated Membrane Protein 2/metabolism , Zebrafish
19.
Methods Enzymol ; 499: 227-58, 2011.
Article in English | MEDLINE | ID: mdl-21683257

ABSTRACT

Transgenic Drosophila melanogaster have been used to model both the physiological and pathological behavior of serpins. The ability to generate flies expressing serpins and to rapidly assess associated phenotypes contributes to the power of this paradigm. While providing a whole-organism model of serpinopathies the powerful toolkit of genetic interventions allows precise molecular dissection of important biological pathways. In this chapter, we summarize the contribution that flies have made to the serpin field and then describe some of the experimental methods that are employed in these studies. In particular, we will describe the generation of transgenic flies, the assessment of phenotypes, and the principles of how to perform a genetic screen.


Subject(s)
Drosophila melanogaster/metabolism , Serpins/metabolism , Animals , Animals, Genetically Modified , Disease Models, Animal , Drosophila melanogaster/genetics , Humans , Protein Conformation , Serpins/genetics
20.
Nat Cell Biol ; 13(4): 453-60, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21394080

ABSTRACT

mTOR (mammalian target of rapamycin) signalling and macroautophagy (henceforth autophagy) regulate numerous pathological and physiological processes, including cellular responses to altered nutrient levels. However, the mechanisms regulating mTOR and autophagy remain incompletely understood. Lysosomes are dynamic intracellular organelles intimately involved both in the activation of mTOR complex 1 (mTORC1) signalling and in degrading autophagic substrates. Here we report that lysosomal positioning coordinates anabolic and catabolic responses with changes in nutrient availability by orchestrating early plasma-membrane signalling events, mTORC1 signalling and autophagy. Activation of mTORC1 by nutrients correlates with its presence on peripheral lysosomes that are physically close to the upstream signalling modules, whereas starvation causes perinuclear clustering of lysosomes, driven by changes in intracellular pH. Lysosomal positioning regulates mTORC1 signalling, which in turn influences autophagosome formation. Lysosome positioning also influences autophagosome-lysosome fusion rates, and thus controls autophagic flux by acting at both the initiation and termination stages of the process. Our findings provide a physiological role for the dynamic state of lysosomal positioning in cells as a coordinator of mTORC1 signalling with autophagic flux.


Subject(s)
Food , Lysosomes/metabolism , Lysosomes/ultrastructure , Proteins/metabolism , Autophagy/physiology , HeLa Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes , Proteins/genetics , Signal Transduction/physiology , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL